51
|
Pablo JL, Greka A. Charting a TRP to Novel Therapeutic Destinations for Kidney Diseases. Trends Pharmacol Sci 2019; 40:911-918. [PMID: 31704171 DOI: 10.1016/j.tips.2019.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/24/2019] [Accepted: 10/07/2019] [Indexed: 12/29/2022]
Abstract
Ion channels are critical to kidney function, and their dysregulation leads to several distinct kidney diseases. Of the diversity of ion channels in kidney cells, the transient receptor potential (TRP) superfamily of proteins plays important and varied roles in both maintaining homeostasis as well as in causing disease. Recent work showed that TRPC5 blockers could successfully protect critical components of the kidney filter both in vitro and in vivo, thus revealing TRPC5 as a tractable therapeutic target for focal and segmental glomerulosclerosis (FSGS), a common cause of kidney failure. Human genetics point to three additional TRP channels as plausible therapeutic targets: TRPC6 in FSGS, PKD2 in polycystic kidney disease, and TRPM6 in familial hypomagnesemia with secondary hypocalcemia (HSH). We conclude that targeting TRP channels could pave the way for much needed therapies for kidney diseases.
Collapse
Affiliation(s)
- Juan Lorenzo Pablo
- Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
52
|
Single-Channel Properties of the ROMK-Pore-Forming Subunit of the Mitochondrial ATP-Sensitive Potassium Channel. Int J Mol Sci 2019; 20:ijms20215323. [PMID: 31731540 PMCID: PMC6862428 DOI: 10.3390/ijms20215323] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
An increased flux of potassium ions into the mitochondrial matrix through the ATP-sensitive potassium channel (mitoKATP) has been shown to provide protection against ischemia-reperfusion injury. Recently, it was proposed that the mitochondrial-targeted isoform of the renal outer medullary potassium channel (ROMK) protein creates a pore-forming subunit of mitoKATP in heart mitochondria. Our research focuses on the properties of mitoKATP from heart-derived H9c2 cells. For the first time, we detected single-channel activity and describe the pharmacology of mitoKATP in the H9c2 heart-derived cells. The patch-clamping of mitoplasts from wild type (WT) and cells overexpressing ROMK2 revealed the existence of a potassium channel that exhibits the same basic properties previously attributed to mitoKATP. ROMK2 overexpression resulted in a significant increase of mitoKATP activity. The conductance of both channels in symmetric 150/150 mM KCl was around 97 ± 2 pS in WT cells and 94 ± 3 pS in cells overexpressing ROMK2. The channels were inhibited by 5-hydroxydecanoic acid (a mitoKATP inhibitor) and by Tertiapin Q (an inhibitor of both the ROMK-type channels and mitoKATP). Additionally, mitoKATP from cells overexpressing ROMK2 were inhibited by ATP/Mg2+ and activated by diazoxide. We used an assay based on proteinase K to examine the topology of the channel in the inner mitochondrial membrane and found that both termini of the protein localized to the mitochondrial matrix. We conclude that the observed activity of the channel formed by the ROMK protein corresponds to the electrophysiological and pharmacological properties of mitoKATP.
Collapse
|
53
|
van der Wijst J, Belge H, Bindels RJM, Devuyst O. Learning Physiology From Inherited Kidney Disorders. Physiol Rev 2019; 99:1575-1653. [PMID: 31215303 DOI: 10.1152/physrev.00008.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The identification of genes causing inherited kidney diseases yielded crucial insights in the molecular basis of disease and improved our understanding of physiological processes that operate in the kidney. Monogenic kidney disorders are caused by mutations in genes coding for a large variety of proteins including receptors, channels and transporters, enzymes, transcription factors, and structural components, operating in specialized cell types that perform highly regulated homeostatic functions. Common variants in some of these genes are also associated with complex traits, as evidenced by genome-wide association studies in the general population. In this review, we discuss how the molecular genetics of inherited disorders affecting different tubular segments of the nephron improved our understanding of various transport processes and of their involvement in homeostasis, while providing novel therapeutic targets. These include inherited disorders causing a dysfunction of the proximal tubule (renal Fanconi syndrome), with emphasis on epithelial differentiation and receptor-mediated endocytosis, or affecting the reabsorption of glucose, the handling of uric acid, and the reabsorption of sodium, calcium, and magnesium along the kidney tubule.
Collapse
Affiliation(s)
- Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Hendrica Belge
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Devuyst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
54
|
Enslow BT, Stockand JD, Berman JM. Liddle's syndrome mechanisms, diagnosis and management. Integr Blood Press Control 2019; 12:13-22. [PMID: 31564964 PMCID: PMC6731958 DOI: 10.2147/ibpc.s188869] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/23/2019] [Indexed: 12/26/2022] Open
Abstract
Liddle's syndrome is a genetic disorder characterized by hypertension with hypokalemic metabolic alkalosis, hyporeninemia and suppressed aldosterone secretion that often appears early in life. It results from inappropriately elevated sodium reabsorption in the distal nephron. Liddle's syndrome is caused by mutations to subunits of the Epithelial Sodium Channel (ENaC). Among other mechanisms, such mutations typically prevent ubiquitination of these subunits, slowing the rate at which they are internalized from the membrane, resulting in an elevation of channel activity. A minority of Liddle's syndrome mutations, though, result in a complementary effect that also elevates activity by increasing the probability that ENaC channels within the membrane are open. Potassium-sparing diuretics such as amiloride and triamterene reduce ENaC activity, and in combination with a reduced sodium diet can restore normotension and electrolyte imbalance in Liddle's syndrome patients and animal models. Liddle's syndrome can be diagnosed clinically by phenotype and confirmed through genetic testing. This review examines the clinical features of Liddle's syndrome, the differential diagnosis of Liddle's syndrome and differentiation from other genetic diseases with similar phenotype, and what is currently known about the population-level prevalence of Liddle's syndrome. This review gives special focus to the molecular mechanisms of Liddle's syndrome.
Collapse
Affiliation(s)
| | | | - Jonathan M Berman
- New York Institute of Technology College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR, USA
| |
Collapse
|
55
|
Li J, Hu S, Nie Y, Wang R, Tan M, Li H, Zhu S. A novel compound heterozygous KCNJ1 gene mutation presenting as late-onset Bartter syndrome: Case report. Medicine (Baltimore) 2019; 98:e16738. [PMID: 31441846 PMCID: PMC6716717 DOI: 10.1097/md.0000000000016738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
RATIONALE Bartter syndrome is an autosomal-recessive inherited disease in which patients present with hypokalemia and metabolic alkalosis. We present 1 case with Bartter syndrome, due to a novel compound heterozygous mutation in the KCNJ1 gene encoding the ATP-sensitive inward rectifier potassium channel in the thick ascending limb of the loop of Henle. PATIENT CONCERNS A patient was admitted to our hospital because of weakness, polyuria, and polydipsia. At presentation to our hospital, the female Chinese patient was 34 years old and her physical examination was normal. Laboratory studies revealed hypokalemia, metabolic alkalosis, hypercalciuria, hyperparathyroidemia, and hyper-reninemia. In addition, urinary potassium was obviously higher. Computer tomography scan confirmed the patient had the bilateral medullary nephrocalcinosis. DIAGNOSIS Blood samples were received from the patient and her parents, and deoxyribonucleic acid was extracted. The genetic analysis of SLC12A1, SLC12A3, KCNJ1, CLCNKB, BSND, and CASR was performed. The compound heterozygous KCNJ1 gene mutation was validated using conventional Sanger sequencing methods. INTERVENTIONS The patient was treated with potassium supplementation. Her blood and urine chemistries improved over the next week. Serum potassium normalized with improvement in polyuria and polydipsia over the next month. OUTCOMES Our patient was compound heterozygous for Thr234Ile and Thr71Met in the KCNJ1 gene. The c.701C>T variant predicted a change from a threonine codon to an isoleucine codon (p.Thr234Ile). The c.212C>T variant predicted a change from a threonine codon to a methionine codon (p.Thr71Met). The unaffected mother was heterozygous for the Thr234Ile mutation, whereas unaffected father was heterozygous for the Thr71Met mutation. LESSONS The phenotypes of the patient were similar to other patients with Bartter syndrome. The phenotypes of the patient could eventually be explained by the presence of the novel compound heterozygous p.Thr234Ile/p.Thr71Met variants in the KCNJ1 gene.
Collapse
Affiliation(s)
- Jingyi Li
- Department of Endocrinology, The First Affiliated Hospital, Tsinghua University
| | - Shoulong Hu
- Department of Ophthalmology, Beijing Children's Hospital, Capital Medical University
| | - Yi Nie
- Department of Endocrinology, The First Affiliated Hospital, Tsinghua University
| | - Rongfeng Wang
- Department of Radiology, The First Affiliated Hospital, Tsinghua University, Beijing, P.R. China
| | - Ming Tan
- Department of Endocrinology, The First Affiliated Hospital, Tsinghua University
| | - Hongmei Li
- Department of Endocrinology, The First Affiliated Hospital, Tsinghua University
| | - Shuanli Zhu
- Department of Endocrinology, The First Affiliated Hospital, Tsinghua University
| |
Collapse
|
56
|
Expression Signatures of Cisplatin- and Trametinib-Treated Early-Stage Medaka Melanomas. G3-GENES GENOMES GENETICS 2019; 9:2267-2276. [PMID: 31101653 PMCID: PMC6643878 DOI: 10.1534/g3.119.400051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Small aquarium fish models provide useful systems not only for a better understanding of the molecular basis of many human diseases, but also for first-line screening to identify new drug candidates. For testing new chemical substances, current strategies mostly rely on easy to perform and efficient embryonic screens. Cancer, however, is a disease that develops mainly during juvenile and adult stage. Long-term treatment and the challenge to monitor changes in tumor phenotype make testing of large chemical libraries in juvenile and adult animals cost prohibitive. We hypothesized that changes in the gene expression profile should occur early during anti-tumor treatment, and the disease-associated transcriptional change should provide a reliable readout that can be utilized to evaluate drug-induced effects. For the current study, we used a previously established medaka melanoma model. As proof of principle, we showed that exposure of melanoma developing fish to the drugs cisplatin or trametinib, known cancer therapies, for a period of seven days is sufficient to detect treatment-induced changes in gene expression. By examining whole body transcriptome responses we provide a novel route toward gene panels that recapitulate anti-tumor outcomes thus allowing a screening of thousands of drugs using a whole-body vertebrate model. Our results suggest that using disease-associated transcriptional change to screen therapeutic molecules in small fish model is viable and may be applied to pre-clinical research and development stages in new drug discovery.
Collapse
|
57
|
Aretz CD, Vadukoot AK, Hopkins CR. Discovery of Small Molecule Renal Outer Medullary Potassium (ROMK) Channel Inhibitors: A Brief History of Medicinal Chemistry Approaches To Develop Novel Diuretic Therapeutics. J Med Chem 2019; 62:8682-8694. [PMID: 31034224 DOI: 10.1021/acs.jmedchem.8b01891] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The renal outer medullary potassium (ROMK) channel is a member of the inwardly rectifying family of potassium (Kir, Kir1.1) channels. It is primarily expressed in two regions of the kidney, the cortical collecting duct (CCD) and the thick ascending loop of Henle (TALH). At the CCD it tightly regulates potassium secretion while controlling potassium recycling in TALH. As loss-of-function mutations lead to salt wasting and low blood pressure, it has been surmised that inhibitors of ROMK would represent a target for new and improved diuretics for the treatment of hypertension and heart failure. In this review, we discuss and provide an overview of the medicinal chemistry approaches toward the development of small molecule ROMK inhibitors over the past decade.
Collapse
Affiliation(s)
- Christopher D Aretz
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Anish K Vadukoot
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| | - Corey R Hopkins
- Department of Pharmaceutical Sciences, College of Pharmacy , University of Nebraska Medical Center , Omaha , Nebraska 68198 , United States
| |
Collapse
|
58
|
Dutter BF, Ender A, Sulikowski GA, Weaver CD. Rhodol-based thallium sensors for cellular imaging of potassium channel activity. Org Biomol Chem 2019; 16:5575-5579. [PMID: 30051127 DOI: 10.1039/c8ob01098f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Thallium (Tl+) flux assays enable imaging of potassium (K+) channel activity in cells and tissues by exploiting the permeability of K+ channels to Tl+ coupled with a fluorescent Tl+ sensitive dye. Common Tl+ sensing dyes utilize fluorescein as the fluorophore though fluorescein exhibits certain undesirable properties in these assays including short excitation wavelengths and pH sensitivity. To overcome these drawbacks, the replacement of fluorescein with rhodols was investigated. A library of 13 rhodol-based Tl+ sensors was synthesized and their properties and performance in Tl+ flux assays evaluated. The dimethyl rhodol Tl+ sensor emerged as the best of the series and performed comparably to fluorescein-based sensors while demonstrating greater pH tolerance in the physiological range and excitation and emission spectra 30 nm red-shifted from fluorescein.
Collapse
Affiliation(s)
- Brendan F Dutter
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.
| | | | | | | |
Collapse
|
59
|
Gonzalez-Vicente A, Saez F, Monzon CM, Asirwatham J, Garvin JL. Thick Ascending Limb Sodium Transport in the Pathogenesis of Hypertension. Physiol Rev 2019; 99:235-309. [PMID: 30354966 DOI: 10.1152/physrev.00055.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The thick ascending limb plays a key role in maintaining water and electrolyte balance. The importance of this segment in regulating blood pressure is evidenced by the effect of loop diuretics or local genetic defects on this parameter. Hormones and factors produced by thick ascending limbs have both autocrine and paracrine effects, which can extend prohypertensive signaling to other structures of the nephron. In this review, we discuss the role of the thick ascending limb in the development of hypertension, not as a sole participant, but one that works within the rich biological context of the renal medulla. We first provide an overview of the basic physiology of the segment and the anatomical considerations necessary to understand its relationship with other renal structures. We explore the physiopathological changes in thick ascending limbs occurring in both genetic and induced animal models of hypertension. We then discuss the racial differences and genetic defects that affect blood pressure in humans through changes in thick ascending limb transport rates. Throughout the text, we scrutinize methodologies and discuss the limitations of research techniques that, when overlooked, can lead investigators to make erroneous conclusions. Thus, in addition to advancing an understanding of the basic mechanisms of physiology, the ultimate goal of this work is to understand our research tools, to make better use of them, and to contextualize research data. Future advances in renal hypertension research will require not only collection of new experimental data, but also integration of our current knowledge.
Collapse
Affiliation(s)
| | - Fara Saez
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Casandra M Monzon
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jessica Asirwatham
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
60
|
Malekshahabi T, Khoshdel Rad N, Serra AL, Moghadasali R. Autosomal dominant polycystic kidney disease: Disrupted pathways and potential therapeutic interventions. J Cell Physiol 2019; 234:12451-12470. [PMID: 30644092 DOI: 10.1002/jcp.28094] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/07/2018] [Indexed: 12/18/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a monogenic inherited renal cystic disease that occurs in different races worldwide. It is characterized by the development of a multitude of renal cysts, which leads to massive enlargement of the kidney and often to renal failure in adulthood. ADPKD is caused by a mutation in PKD1 or PKD2 genes encoding the proteins polycystin-1 and polycystin-2, respectively. Recent studies showed that cyst formation and growth result from deregulation of multiple cellular pathways like proliferation, apoptosis, metabolic processes, cell polarity, and immune defense. In ADPKD, intracellular cyclic adenosine monophosphate (cAMP) promotes cyst enlargement by stimulating cell proliferation and transepithelial fluid secretion. Several interventions affecting many of these defective signaling pathways have been effective in animal models and some are currently being tested in clinical trials. Moreover, the stem cell therapy can improve nephropathies and according to studies were done in this field, can be considered as a hopeful therapeutic approach in future for PKD. This study provides an in-depth review of the relevant molecular pathways associated with the pathogenesis of ADPKD and their implications in development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Talieh Malekshahabi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Andreas L Serra
- Department of Internal Medicine and Nephrology, Klinik Hirslanden, Zurich, Switzerland
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
61
|
Olejnik A, Franczak A, Krzywonos-Zawadzka A, Kałużna-Oleksy M, Bil-Lula I. The Biological Role of Klotho Protein in the Development of Cardiovascular Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5171945. [PMID: 30671457 PMCID: PMC6323445 DOI: 10.1155/2018/5171945] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/09/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022]
Abstract
Klotho is a membrane-bound or soluble antiaging protein, whose protective activity is essential for a proper function of many organs. In 1997, an accidental insertion of a transgene led to creation of transgenic mice with several age-related disorders. In Klotho-deficient mice, the inherited phenotypes closely resemble human aging, while in an animal model of Klotho overexpression, the lifespan is extended. Klotho protein is detected mainly in the kidneys and brain. It is a coreceptor for fibroblast growth factor and hence is involved in maintaining endocrine system homeostasis. Furthermore, an inhibition of insulin/insulin-like growth factor-1 signaling pathway by Klotho regulates oxidative stress and reduces cell death. The association between serum Klotho and the classic risk factors, as well as the clinical history of cardiovascular disease, was also shown. There are a lot of evidences that Klotho deficiency correlates with the occurrence and development of coronary artery disease, atherosclerosis, myocardial infarction, and left ventricular hypertrophy. Therefore, an involvement of Klotho in the signaling pathways and in regulation of a proper cell metabolism could be a crucial factor in the cardiac and vascular protection. It is also well established that Klotho protein enhances the antioxidative response via augmented production of superoxide dismutase and reduced generation of reactive oxygen species. Recent studies have proven an expression of Klotho in cardiomyocytes and its increased expression in stress-related heart injury. Thus, the antioxidative and antiapoptotic activity of Klotho could be considered as the novel protective factor in cardiovascular disease and heart injury.
Collapse
Affiliation(s)
- Agnieszka Olejnik
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Aleksandra Franczak
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Anna Krzywonos-Zawadzka
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Marta Kałużna-Oleksy
- Department of Cardiology, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, 61-848 Poznan, Poland
| | - Iwona Bil-Lula
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
62
|
Teulon J, Planelles G, Sepúlveda FV, Andrini O, Lourdel S, Paulais M. Renal Chloride Channels in Relation to Sodium Chloride Transport. Compr Physiol 2018; 9:301-342. [DOI: 10.1002/cphy.c180024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
63
|
Kamel KS, Schreiber M, Halperin ML. Renal potassium physiology: integration of the renal response to dietary potassium depletion. Kidney Int 2018; 93:41-53. [PMID: 29102372 DOI: 10.1016/j.kint.2017.08.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/31/2017] [Accepted: 08/03/2017] [Indexed: 01/30/2023]
Abstract
We summarize the current understanding of the physiology of the renal handling of potassium (K+), and present an integrative view of the renal response to K+ depletion caused by dietary K+ restriction. This renal response involves contributions from different nephron segments, and aims to diminish the rate of excretion of K+ as a result of: decreasing the rate of electrogenic (and increasing the rate of electroneutral) reabsorption of sodium in the aldosterone-sensitive distal nephron (ASDN), decreasing the abundance of renal outer medullary K+ channels in the luminal membrane of principal cells in the ASDN, decreasing the flow rate in the ASDN, and increasing the reabsorption of K+ in the cortical and medullary collecting ducts. The implications of this physiology for the association between K+ depletion and hypertension, and K+ depletion and formation of calcium kidney stones are discussed.
Collapse
Affiliation(s)
- Kamel S Kamel
- Renal Division, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.
| | - Martin Schreiber
- Renal Division, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mitchell L Halperin
- Renal Division, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
64
|
Abstract
Bartter syndrome is an inherited renal tubular disorder caused by a defective salt reabsorption in the thick ascending limb of loop of Henle, resulting in salt wasting, hypokalemia, and metabolic alkalosis. Mutations of several genes encoding the transporters and channels involved in salt reabsorption in the thick ascending limb cause different types of Bartter syndrome. A poor phenotype-genotype relationship due to the interaction with other cotransporters and different degrees of compensation through alternative pathways is currently reported. However, phenotypic identification still remains the first step to guide the suspicion of Bartter syndrome. Given the rarity of the syndrome, and the lack of genetic characterization in most cases, limited clinical evidence for treatment is available and the therapy is based mainly on the comprehension of renal physiology and relies on the physician's personal experiences. A better understanding of the mutated channels and transporters could possibly generate targets for specific treatment in the future, also encompassing drugs aiming to correct deficiencies in folding or plasma membrane expression of the mutated proteins.
Collapse
Affiliation(s)
- Tamara da Silva Cunha
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, São Paulo, Brazil,
| | - Ita Pfeferman Heilberg
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, São Paulo, Brazil,
| |
Collapse
|
65
|
Otterpohl KL, Hart RG, Evans C, Surendran K, Chandrasekar I. Nonmuscle myosin 2 proteins encoded by Myh9, Myh10, and Myh14 are uniquely distributed in the tubular segments of murine kidney. Physiol Rep 2018; 5. [PMID: 29208685 PMCID: PMC5727274 DOI: 10.14814/phy2.13513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 11/24/2022] Open
Abstract
The diverse epithelial cell types of the kidneys are segregated into nephron segments and the collecting ducts in order to endow each tubular segment with unique functions. The rich diversity of the epithelial cell types is highlighted by the unique membrane channels and receptors expressed within each nephron segment. Our previous work identified a critical role for Myh9 and Myh10 in mammalian endocytosis. Here, we examined the expression patterns of Nonmuscle myosin 2 (NM2) heavy chains encoded by Myh9, Myh10, and Myh14 in mouse kidneys as these genes may confer unique nephron segment‐specific membrane transport properties. Interestingly, we found that each segment of the renal tubules predominately expressed only two of the three NM2 isoforms, with isoform‐specific subcellular localization, and different levels of expression within a nephron segment. Additionally, we identify Myh14 to be restricted to the intercalated cells and Myh10 to be restricted to the principal cells within the collecting ducts and connecting segments. We speculate that the distinct expression pattern of the NM2 proteins likely reflects the diversity of the intracellular trafficking machinery present within the different renal tubular epithelial segments.
Collapse
Affiliation(s)
- Karla L Otterpohl
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA
| | - Ryan G Hart
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA
| | - Claire Evans
- Molecular Pathology Core, Sanford Research, Sioux Falls, South Dakota, USA
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group - Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, South Dakota, USA
| | - Indra Chandrasekar
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, South Dakota, USA
| |
Collapse
|
66
|
Ramratnam M, Kenny B, Kyle JW, Wiedmeyer B, Hacker TA, Barefield DY, McNally EM, Makielski JC. Transgenic overexpression of the SUR2A-55 splice variant in mouse heart reduces infract size and promotes protective mitochondrial function. Heliyon 2018; 4:e00677. [PMID: 29998196 PMCID: PMC6037880 DOI: 10.1016/j.heliyon.2018.e00677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/28/2018] [Indexed: 01/21/2023] Open
Abstract
ATP-sensitive potassium channels found in both the sarcolemma (sarcKATP) and mitochondria (mitoKATP) of cardiomyocytes are important mediators of cardioprotection during ischemic heart disease. Sulfonylurea receptor isoforms (SUR2), encoded by Abcc9, an ATP-binding cassette family member, form regulatory subunits of the sarcKATP channel and are also thought to regulate mitoKATP channel activity. A short-form splice variant of SUR2 (SUR2A-55) was previously shown to target mitochondria and display diaxoxide and ATP insensitive KATP activity when co-expressed with the inward rectifier channels Kir6.2 and Kir6.1. We hypothesized that mice with cardiac specific overexpression of SUR2A-55 would mediate cardioprotection from ischemia by altering mitoKATP properties. Mice overexpressing SUR2A-55 (TGSUR2A-55) in cardiomyocytes were generated and showed no significant difference in echocardiographic measured chamber dimension, percent fractional shortening, heart to body weight ratio, or gross histologic features compared to normal mice at 11–14 weeks of age. TGSUR2A-55 had improved hemodynamic functional recovery and smaller infarct size after ischemia reperfusion injury compared to WT mice in an isolated hanging heart model. The mitochondrial membrane potential of TGSUR2A-55 mice was less sensitive to ATP, diazoxide, and Ca2+ loading. These data suggest that the SUR2A-55 splice variant favorably affects mitochondrial function leading to cardioprotection. These data support a role for the regulation of mitoKATP activity by SUR2A-55.
Collapse
Affiliation(s)
- Mohun Ramratnam
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Cardiology Section, Medical Service, William. S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Barrett Kenny
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - John W Kyle
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Brandi Wiedmeyer
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Timothy A Hacker
- Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - David Y Barefield
- Center for Genetic Medicine, Northwestern University, Chicago, IL, United States
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University, Chicago, IL, United States
| | - Jonathan C Makielski
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
67
|
Mackie TD, Brodsky JL. Investigating Potassium Channels in Budding Yeast: A Genetic Sandbox. Genetics 2018; 209:637-650. [PMID: 29967058 PMCID: PMC6028241 DOI: 10.1534/genetics.118.301026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/15/2018] [Indexed: 12/26/2022] Open
Abstract
Like all species, the model eukaryote Saccharomyces cerevisiae, or Bakers' yeast, concentrates potassium in the cytosol as an electrogenic osmolyte and enzyme cofactor. Yeast are capable of robust growth on a wide variety of potassium concentrations, ranging from 10 µM to 2.5 M, due to the presence of a high-affinity potassium uptake system and a battery of cation exchange transporters. Genetic perturbation of either of these systems retards yeast growth on low or high potassium, respectively. However, these potassium-sensitized yeast are a powerful genetic tool, which has been leveraged for diverse studies. Notably, the potassium-sensitive cells can be transformed with plasmids encoding potassium channels from bacteria, plants, or mammals, and subsequent changes in growth rate have been found to correlate with the activity of the introduced potassium channel. Discoveries arising from the use of this assay over the past three decades have increased our understanding of the structure-function relationships of various potassium channels, the mechanisms underlying the regulation of potassium channel function and trafficking, and the chemical basis of potassium channel modulation. In this article, we provide an overview of the major genetic tools used to study potassium channels in S. cerevisiae, a survey of seminal studies utilizing these tools, and a prospective for the future use of this elegant genetic approach.
Collapse
Affiliation(s)
- Timothy D Mackie
- Department of Biological Sciences, University of Pittsburgh, Pennsylvania 15260
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pennsylvania 15260
| |
Collapse
|
68
|
Mackie TD, Kim BY, Subramanya AR, Bain DJ, O'Donnell AF, Welling PA, Brodsky JL. The endosomal trafficking factors CORVET and ESCRT suppress plasma membrane residence of the renal outer medullary potassium channel (ROMK). J Biol Chem 2018; 293:3201-3217. [PMID: 29311259 PMCID: PMC5836112 DOI: 10.1074/jbc.m117.819086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 01/02/2018] [Indexed: 11/06/2022] Open
Abstract
Protein trafficking can act as the primary regulatory mechanism for ion channels with high open probabilities, such as the renal outer medullary (ROMK) channel. ROMK, also known as Kir1.1 (KCNJ1), is the major route for potassium secretion into the pro-urine and plays an indispensable role in regulating serum potassium and urinary concentrations. However, the cellular machinery that regulates ROMK trafficking has not been fully defined. To identify regulators of the cell-surface population of ROMK, we expressed a pH-insensitive version of the channel in the budding yeast Saccharomyces cerevisiae We determined that ROMK primarily resides in the endoplasmic reticulum (ER), as it does in mammalian cells, and is subject to ER-associated degradation (ERAD). However, sufficient ROMK levels on the plasma membrane rescued growth on low-potassium medium of yeast cells lacking endogenous potassium channels. Next, we aimed to identify the biological pathways most important for ROMK regulation. Therefore, we used a synthetic genetic array to identify non-essential genes that reduce the plasma membrane pool of ROMK in potassium-sensitive yeast cells. Genes identified in this screen included several members of the endosomal complexes required for transport (ESCRT) and the class-C core vacuole/endosome tethering (CORVET) complexes. Mass spectroscopy analysis confirmed that yeast cells lacking an ESCRT component accumulate higher potassium concentrations. Moreover, silencing of ESCRT and CORVET components increased ROMK levels at the plasma membrane in HEK293 cells. Our results indicate that components of the post-endocytic pathway influence the cell-surface density of ROMK and establish that components in this pathway modulate channel activity.
Collapse
Affiliation(s)
| | - Bo-Young Kim
- the Department of Physiology, University of Maryland at Baltimore, Baltimore, Maryland 21201
| | - Arohan R Subramanya
- the Departments of Medicine and Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- the Medicine and Research Services, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania 15240, and
| | - Daniel J Bain
- Geology and Environmental Science, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Allyson F O'Donnell
- the Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania 15282
| | - Paul A Welling
- the Department of Physiology, University of Maryland at Baltimore, Baltimore, Maryland 21201
| | | |
Collapse
|
69
|
Bednarczyk P, Kicinska A, Laskowski M, Kulawiak B, Kampa R, Walewska A, Krajewska M, Jarmuszkiewicz W, Szewczyk A. Evidence for a mitochondrial ATP-regulated potassium channel in human dermal fibroblasts. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:309-318. [PMID: 29458000 DOI: 10.1016/j.bbabio.2018.02.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/04/2018] [Accepted: 02/14/2018] [Indexed: 02/07/2023]
Abstract
Mitochondrial ATP-regulated potassium channels are present in the inner membrane of the mitochondria of various cells. In the present study, we show for the first time mitochondrial ATP-regulated potassium channels in human dermal fibroblast cells. Using the patch-clamp technique on the inner mitochondrial membrane of fibroblasts, we detected a potassium channel with a mean conductance equal to 100 pS in symmetric 150 mM KCl. The activity of this channel was inhibited by a complex of ATP/Mg2+ and activated by potassium channel openers such as diazoxide or BMS 191095. Channel activity was inhibited by antidiabetic sulfonylurea glibenclamide and 5-hydroxydecanoic acid. The influence of substances modulating ATP-regulated potassium channel activity on oxygen consumption and membrane potential of isolated fibroblast mitochondria was also studied. Additionally, the potassium channel opener diazoxide lowered the amount of superoxide formed in isolated fibroblast mitochondria. Using reverse transcriptase-PCR, we found an mRNA transcript for the KCNJ1(ROMK) channel. The presence of ROMK protein was observed in the inner mitochondrial membrane fraction. Moreover, colocalization of the ROMK protein and a mitochondrial marker in the mitochondria of fibroblast cells was shown by immunofluorescence. In summary, the ATP-regulated mitochondrial potassium channel in a dermal fibroblast cell line have been identified.
Collapse
Affiliation(s)
- Piotr Bednarczyk
- Department of Biophysics, Warsaw University of Life Sciences (SGGW), Warsaw, Poland.
| | - Anna Kicinska
- Department of Bioenergetics, Adam Mickiewicz University, Poznan, Poland
| | - Michal Laskowski
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bogusz Kulawiak
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Rafal Kampa
- Department of Biophysics, Warsaw University of Life Sciences (SGGW), Warsaw, Poland; Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Agnieszka Walewska
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Milena Krajewska
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | - Adam Szewczyk
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
70
|
Sammons MF, Kharade SV, Filipski KJ, Boehm M, Smith AC, Shavnya A, Fernando DP, Dowling MS, Carpino PA, Castle NA, Zellmer SG, Antonio BM, Gosset JR, Carlo A, Denton JS. Discovery and in Vitro Optimization of 3-Sulfamoylbenzamides as ROMK Inhibitors. ACS Med Chem Lett 2018; 9:125-130. [PMID: 29456800 DOI: 10.1021/acsmedchemlett.7b00481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/19/2018] [Indexed: 02/05/2023] Open
Abstract
Inhibitors of the renal outer medullary potassium channel (ROMK) show promise as novel mechanism diuretics, with potentially lower risk of diuretic-induced hypokalemia relative to current thiazide and loop diuretics. Here, we report the identification of a novel series of 3-sulfamoylbenzamide ROMK inhibitors. Starting from HTS hit 4, this series was optimized to provide ROMK inhibitors with good in vitro potencies and well-balanced ADME profiles. In contrast to previously reported small-molecule ROMK inhibitors, members of this series were demonstrated to be highly selective for inhibition of human over rat ROMK and to be insensitive to the N171D pore mutation that abolishes inhibitory activity of previously reported ROMK inhibitors.
Collapse
Affiliation(s)
- Matthew F. Sammons
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Sujay V. Kharade
- Department
of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Kevin J. Filipski
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Markus Boehm
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Aaron C. Smith
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Andre Shavnya
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Dilinie P. Fernando
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew S. Dowling
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Philip A. Carpino
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Neil A. Castle
- Neusentis, Pfizer Worldwide Research & Development, Durham, North Carolina 27703, United States
| | - Shannon G. Zellmer
- Neusentis, Pfizer Worldwide Research & Development, Durham, North Carolina 27703, United States
| | - Brett M. Antonio
- Neusentis, Pfizer Worldwide Research & Development, Durham, North Carolina 27703, United States
| | - James R. Gosset
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Anthony Carlo
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jerod S. Denton
- Department
of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
71
|
Potassium Regulation in Medaka (Oryzias latipes) Larvae Acclimated to Fresh Water: Passive Uptake and Active Secretion by the Skin Cells. Sci Rep 2017; 7:16215. [PMID: 29176723 PMCID: PMC5701230 DOI: 10.1038/s41598-017-16381-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/13/2017] [Indexed: 01/29/2023] Open
Abstract
Molecular mechanisms of Na+, Cl−, and Ca2+ regulation in ionocytes of fish have been well investigated. However, the regulatory mechanism of K+ in fishes has been largely unknown. In this study, we investigated the mechanism of K+ regulation in medaka larvae acclimated to fresh water. Using a scanning ion-selective electrode technique (SIET) to measure the K+ fluxes at skin cells, significant K+ effluxes were found at ionocytes; in contrast, significant K+ influxes were found at the boundaries between keratinocytes. High K+ water (HK) acclimation induced the K+ effluxes at ionocytes and suppressed the K+ influxes at keratinocytes. The K+ effluxes of ionocytes were suppressed by VU591, bumetanide and ouabain. The K+ influxes of keratinocytes were suppressed by TAP. In situ hybridization analysis showed that mRNA of ROMKa was expressed by ionocytes in the skin and gills of medaka larvae. Quantitative PCR showed that mRNA levels of ROMKa and NKCC1a in gills of adult medaka were upregulated after HK acclimation. This study suggests that medaka obtain K+ through a paracellular pathway between keratinocytes and extrude K+ through ionocytes; apical ROMKa and basolateral NKCC1a are involved in the K+ secretion by ionocytes.
Collapse
|
72
|
Kharade SV, Sheehan JH, Figueroa EE, Meiler J, Denton JS. Pore Polarity and Charge Determine Differential Block of Kir1.1 and Kir7.1 Potassium Channels by Small-Molecule Inhibitor VU590. Mol Pharmacol 2017; 92:338-346. [PMID: 28619748 PMCID: PMC5553192 DOI: 10.1124/mol.117.108472] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/12/2017] [Indexed: 12/28/2022] Open
Abstract
VU590 was the first publicly disclosed, submicromolar-affinity (IC50 = 0.2 μM), small-molecule inhibitor of the inward rectifier potassium (Kir) channel and diuretic target, Kir1.1. VU590 also inhibits Kir7.1 (IC50 ∼ 8 μM), and has been used to reveal new roles for Kir7.1 in regulation of myometrial contractility and melanocortin signaling. Here, we employed molecular modeling, mutagenesis, and patch clamp electrophysiology to elucidate the molecular mechanisms underlying VU590 inhibition of Kir1.1 and Kir7.1. Block of both channels is voltage- and K+-dependent, suggesting the VU590 binding site is located within the pore. Mutagenesis analysis in Kir1.1 revealed that asparagine 171 (N171) is the only pore-lining residue required for high-affinity block, and that substituting negatively charged residues (N171D, N171E) at this position dramatically weakens block. In contrast, substituting a negatively charged residue at the equivalent position in Kir7.1 enhances block by VU590, suggesting the VU590 binding mode is different. Interestingly, mutations of threonine 153 (T153) in Kir7.1 that reduce constrained polarity at this site (T153C, T153V, T153S) make wild-type and binding-site mutants (E149Q, A150S) more sensitive to block by VU590. The Kir7.1-T153C mutation enhances block by the structurally unrelated inhibitor VU714 but not by a higher-affinity analog ML418, suggesting that the polar side chain of T153 creates a barrier to low-affinity ligands that interact with E149 and A150. Reverse mutations in Kir1.1 suggest that this mechanism is conserved in other Kir channels. This study reveals a previously unappreciated role of membrane pore polarity in determination of Kir channel inhibitor pharmacology.
Collapse
Affiliation(s)
- Sujay V Kharade
- Department of Anesthesiology (S.V.K., E.E.F., J.S.D.), Department of Pharmacology (E.E.F., J.S.D.), Department of Biochemistry (J.H.S., J.M.), Center for Structural Biology (J.H.S., J.M.), Department of Chemistry (J.M.), Institute of Chemical Biology (J.S.D.), Vanderbilt University, Nashville, Tennessee
| | - Jonathan H Sheehan
- Department of Anesthesiology (S.V.K., E.E.F., J.S.D.), Department of Pharmacology (E.E.F., J.S.D.), Department of Biochemistry (J.H.S., J.M.), Center for Structural Biology (J.H.S., J.M.), Department of Chemistry (J.M.), Institute of Chemical Biology (J.S.D.), Vanderbilt University, Nashville, Tennessee
| | - Eric E Figueroa
- Department of Anesthesiology (S.V.K., E.E.F., J.S.D.), Department of Pharmacology (E.E.F., J.S.D.), Department of Biochemistry (J.H.S., J.M.), Center for Structural Biology (J.H.S., J.M.), Department of Chemistry (J.M.), Institute of Chemical Biology (J.S.D.), Vanderbilt University, Nashville, Tennessee
| | - Jens Meiler
- Department of Anesthesiology (S.V.K., E.E.F., J.S.D.), Department of Pharmacology (E.E.F., J.S.D.), Department of Biochemistry (J.H.S., J.M.), Center for Structural Biology (J.H.S., J.M.), Department of Chemistry (J.M.), Institute of Chemical Biology (J.S.D.), Vanderbilt University, Nashville, Tennessee
| | - Jerod S Denton
- Department of Anesthesiology (S.V.K., E.E.F., J.S.D.), Department of Pharmacology (E.E.F., J.S.D.), Department of Biochemistry (J.H.S., J.M.), Center for Structural Biology (J.H.S., J.M.), Department of Chemistry (J.M.), Institute of Chemical Biology (J.S.D.), Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
73
|
Wang B, Wen D, Li H, Wang-France J, Sansom SC. Net K + secretion in the thick ascending limb of mice on a low-Na, high-K diet. Kidney Int 2017; 92:864-875. [PMID: 28688582 DOI: 10.1016/j.kint.2017.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/23/2017] [Accepted: 04/06/2017] [Indexed: 12/29/2022]
Abstract
Because of its cardio-protective effects, a low-Na, high-K diet (LNaHK) is often warranted in conjunction with diuretics to treat hypertensive patients. However, it is necessary to understand the renal handling of such diets in order to choose the best diuretic. Wild-type (WT) or Renal Outer Medullary K channel (ROMK) knockout mice (KO) were given a regular (CTRL), LNaHK, or high-K diet (HK) for 4-7 days. On LNaHK, mice treated with either IP furosemide for 12 hrs, or given furosemide in drinking water for 7 days, exhibited decreased K clearance. We used free-flow micropuncture to measure the [K+] in the early distal tubule (EDT [K+]) before and after furosemide treatment. Furosemide increased the EDT [K+] in WT on CTRL but decreased that in WT on LNaHK. Furosemide did not affect the EDT [K+] of KO on LNaHK or WT on HK. Furosemide-sensitive Na+ excretion was significantly greater in mice on LNaHK than those on CTRL or HK. Patch clamp analysis of split-open TALs revealed that 70-pS ROMK exhibited a higher open probability (Po) but similar density in mice on LNaHK, compared with CTRL. No difference was found in the density or Po of the 30 pS K channels between the two groups. These results indicate mice on LNaHK exhibited furosemide-sensitive net K+ secretion in the TAL that is dependent on increased NKCC2 activity and mediated by ROMK. We conclude that furosemide is a K-sparing diuretic by decreasing the TAL net K+ secretion in subjects on LNaHK.
Collapse
Affiliation(s)
- Bangchen Wang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Donghai Wen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Huaqing Li
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jun Wang-France
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Steven C Sansom
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
74
|
O'Donnell BM, Mackie TD, Subramanya AR, Brodsky JL. Endoplasmic reticulum-associated degradation of the renal potassium channel, ROMK, leads to type II Bartter syndrome. J Biol Chem 2017. [PMID: 28630040 DOI: 10.1074/jbc.m117.786376] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Type II Bartter syndrome is caused by mutations in the renal outer medullary potassium (ROMK) channel, but the molecular mechanisms underlying this disease are poorly defined. To rapidly screen for ROMK function, we developed a yeast expression system and discovered that yeast cells lacking endogenous potassium channels could be rescued by WT ROMK but not by ROMK proteins containing any one of four Bartter mutations. We also found that the mutant proteins were significantly less stable than WT ROMK. However, their degradation was slowed in the presence of a proteasome inhibitor or when yeast cells contained mutations in the CDC48 or SSA1 gene, which is required for endoplasmic reticulum (ER)-associated degradation (ERAD). Consistent with these data, sucrose gradient centrifugation and indirect immunofluorescence microscopy indicated that most ROMK protein was ER-localized. To translate these findings to a more relevant cell type, we measured the stabilities of WT ROMK and the ROMK Bartter mutants in HEK293 cells. As in yeast, the Bartter mutant proteins were less stable than the WT protein, and their degradation was slowed in the presence of a proteasome inhibitor. Finally, we discovered that low-temperature incubation increased the steady-state levels of a Bartter mutant, suggesting that the disease-causing mutation traps the protein in a folding-deficient conformation. These findings indicate that the underlying pathology for at least a subset of patients with type II Bartter syndrome is linked to the ERAD pathway and that future therapeutic strategies should focus on correcting deficiencies in ROMK folding.
Collapse
Affiliation(s)
- Brighid M O'Donnell
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260; Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Timothy D Mackie
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Arohan R Subramanya
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.
| |
Collapse
|
75
|
Zhang X, Wen H, Wang H, Ren Y, Zhao J, Li Y. RNA-Seq analysis of salinity stress-responsive transcriptome in the liver of spotted sea bass (Lateolabrax maculatus). PLoS One 2017; 12:e0173238. [PMID: 28253338 PMCID: PMC5333887 DOI: 10.1371/journal.pone.0173238] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/18/2017] [Indexed: 12/16/2022] Open
Abstract
Salinity is one of the most prominent abiotic factors, which greatly influence reproduction, development, growth, physiological and metabolic activities of fishes. Spotted sea bass (Lateolabrax maculatus), as a euryhaline marine teleost, has extraordinary ability to deal with a wide range of salinity changes. However, this species is devoid of genomic resources, and no study has been conducted at the transcriptomic level to determine genes responsible for salinity regulation, which impedes the understanding of the fundamental mechanism conferring tolerance to salinity fluctuations. Liver, as the major metabolic organ, is the key source supplying energy for iono- and osmoregulation in fish, however, little attention has been paid to its salinity-related functions but which should not be ignored. In this study, we perform RNA-Seq analysis to identify genes involved in salinity adaptation and osmoregulation in liver of spotted sea bass, generating from the fishes exposed to low and high salinity water (5 vs 30ppt). After de novo assembly, annotation and differential gene expression analysis, a total of 455 genes were differentially expressed, including 184 up-regulated and 271 down-regulated transcripts in low salinity-acclimated fish group compared with that in high salinity-acclimated group. A number of genes with a potential role in salinity adaptation for spotted sea bass were classified into five functional categories based on the gene ontology (GO) and enrichment analysis, which include genes involved in metabolites and ion transporters, energy metabolism, signal transduction, immune response and structure reorganization. The candidate genes identified in L. maculates liver provide valuable information to explore new pathways related to fish salinity and osmotic regulation. Besides, the transcriptomic sequencing data supplies significant resources for identification of novel genes and further studying biological questions in spotted sea bass.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- The Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Haishen Wen
- The Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Hailiang Wang
- The Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Yuanyuan Ren
- The Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Ji Zhao
- The Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| | - Yun Li
- The Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Ocean University of China, Qingdao, P. R. China
| |
Collapse
|
76
|
Lee JD, Lee MH, Yang WK, Wang KL, Lee TH. Differential Expression of Renal Outer Medullary K + Channel and Voltage-gated K + Channel 7.1 in Bladder Urothelium of Patients With Interstitial Cystitis/Painful Bladder Syndrome. Urology 2017; 101:169.e1-169.e5. [DOI: 10.1016/j.urology.2016.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 10/04/2016] [Accepted: 11/04/2016] [Indexed: 12/27/2022]
|
77
|
Araujo M, Welch WJ, Zhou X, Sullivan K, Walsh S, Pasternak A, Wilcox CS. Inhibition of ROMK blocks macula densa tubuloglomerular feedback yet causes renal vasoconstriction in anesthetized rats. Am J Physiol Renal Physiol 2017; 312:F1120-F1127. [PMID: 28228405 DOI: 10.1152/ajprenal.00662.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 11/22/2022] Open
Abstract
The Na+-K+-2Cl- cotransporter (NKCC2) on the loop of Henle is the site of action of furosemide. Because outer medullary potassium channel (ROMK) inhibitors prevent reabsorption by NKCC2, we tested the hypothesis that ROMK inhibition with a novel selective ROMK inhibitor (compound C) blocks tubuloglomerular feedback (TGF) and reduces vascular resistance. Loop perfusion of either ROMK inhibitor or furosemide caused dose-dependent blunting of TGF, but the response to furosemide was 10-fold more sensitive (IC50 = 10-6 M for furosemide and IC50 = 10-5 M for compound C). During systemic infusion, both diuretics inhibited TGF, but ROMK inhibitor was 10-fold more sensitive (compound C: 63% inhibition; furosemide: 32% inhibition). Despite blockade of TGF, 1 h of constant systemic infusion of both diuretics reduced the glomerular filtration rate (GFR) and renal blood flow (RBF) by 40-60% and increased renal vascular resistance (RVR) by 100-200%. Neither diuretic altered blood pressure or hematocrit. Proximal tubule hydrostatic pressures (PPT) increased transiently with both diuretics (compound C: 56% increase; furosemide: 70% increase) but returned to baseline. ROMK inhibitor caused more natriuresis (3,400 vs. 1,600% increase) and calciuresis (1,200 vs. 800% increase) but less kaliuresis (33 vs. 167% increase) than furosemide. In conclusion, blockade of ROMK or Na+-K+-2Cl- transport inhibits TGF yet increases renal vascular resistance. The renal vasoconstriction was independent of volume depletion, blood pressure, TGF, or PPT.
Collapse
Affiliation(s)
- Magali Araujo
- Hypertension Research Center and Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia; and
| | - William J Welch
- Hypertension Research Center and Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia; and
| | - Xiaoyan Zhou
- Department of Cardiometabolic Diseases, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Kathleen Sullivan
- Department of Cardiometabolic Diseases, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Shawn Walsh
- Department of Cardiometabolic Diseases, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Alexander Pasternak
- Department of Cardiometabolic Diseases, Merck & Company, Incorporated, Kenilworth, New Jersey
| | - Christopher S Wilcox
- Hypertension Research Center and Division of Nephrology and Hypertension, Georgetown University, Washington, District of Columbia; and
| |
Collapse
|
78
|
Zhou X, Forrest MJ, Sharif-Rodriguez W, Forrest G, Szeto D, Urosevic-Price O, Zhu Y, Stevenson AS, Zhou Y, Stribling S, Dajee M, Walsh SP, Pasternak A, Sullivan KA. Chronic Inhibition of Renal Outer Medullary Potassium Channel Not Only Prevented but Also Reversed Development of Hypertension and End-Organ Damage in Dahl Salt-Sensitive Rats. Hypertension 2017; 69:332-338. [DOI: 10.1161/hypertensionaha.116.08358] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 09/05/2016] [Accepted: 11/13/2016] [Indexed: 01/21/2023]
Abstract
The renal outer medullary potassium (ROMK) channel mediates potassium recycling and facilitates sodium reabsorption through the Na
+
/K
+
/2Cl
−
cotransporter in the loop of Henle and potassium secretion at the cortical collecting duct. Evidence from the phenotype of humans and rodents with functional ROMK deficiency supports the contention that selective ROMK inhibitors (ROMKi) will represent a novel diuretic with potential of therapeutic benefit for hypertension. ROMKi have recently been synthesized by Merck & Co, Inc. The present studies were designed to examine the effects of ROMKi B on systemic hemodynamics, renal function and structure, and vascular function in Dahl salt-sensitive rats. Four experimental groups—control, high-salt diet alone; ROMKi B 3 mg·kg
−
1
·d
−
1
; ROMKi B 10 mg·kg
−
1
·d
−
1
; and hydrochlorothiazide 25 mg·kg
−
1
·d
−
1
—were included in prophylactic (from week 1 to week 9 on high-salt diet) and therapeutic studies (from week 5 to week 9 on high-salt diet), respectively. ROMKi B produced sustained blood pressure reduction and improved renal and vascular function and histological alterations induced by a high-salt diet. ROMKi B was superior to hydrochlorothiazide at reducing blood pressure. Furthermore, ROMKi B provided beneficial effects on both the plasma lipid profile and bone mineral density. Chronic ROMK inhibition not only prevented but also reversed the development of hypertension and end-organ damage in Dahl salt-sensitive rats. Our findings suggest a potential utility of ROMKi B as a novel antihypertensive agent, particularly for the treatment of the salt-sensitive hypertension patient population.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Michael J. Forrest
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Wanda Sharif-Rodriguez
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Gail Forrest
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Daphne Szeto
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Olga Urosevic-Price
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Yonghua Zhu
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Andra S. Stevenson
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Yuchen Zhou
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Sloan Stribling
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Maya Dajee
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Shawn P. Walsh
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Alexander Pasternak
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| | - Kathleen A. Sullivan
- From the Departments of Cardiometabolic Diseases (X.Z., W.S.-R., Y.Z., A.S.S., M.D., K.A.S.), In Vivo Pharmacology (M.J.F., G.F., D.S., O.U.-P., Y.Z., S.S.), and Chemistry (S.P.W., A.P.), Merck & Co, Inc, Kenilworth, NJ
| |
Collapse
|
79
|
Abdel-Magid AF. Potential of Renal Outer Medullary Potassium (ROMK) Channel as Treatments for Hypertension and Heart Failure. ACS Med Chem Lett 2017; 8:9-10. [PMID: 28105264 DOI: 10.1021/acsmedchemlett.6b00457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Indexed: 11/30/2022] Open
|
80
|
Imbrici P, Liantonio A, Camerino GM, De Bellis M, Camerino C, Mele A, Giustino A, Pierno S, De Luca A, Tricarico D, Desaphy JF, Conte D. Therapeutic Approaches to Genetic Ion Channelopathies and Perspectives in Drug Discovery. Front Pharmacol 2016; 7:121. [PMID: 27242528 PMCID: PMC4861771 DOI: 10.3389/fphar.2016.00121] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 04/25/2016] [Indexed: 12/21/2022] Open
Abstract
In the human genome more than 400 genes encode ion channels, which are transmembrane proteins mediating ion fluxes across membranes. Being expressed in all cell types, they are involved in almost all physiological processes, including sense perception, neurotransmission, muscle contraction, secretion, immune response, cell proliferation, and differentiation. Due to the widespread tissue distribution of ion channels and their physiological functions, mutations in genes encoding ion channel subunits, or their interacting proteins, are responsible for inherited ion channelopathies. These diseases can range from common to very rare disorders and their severity can be mild, disabling, or life-threatening. In spite of this, ion channels are the primary target of only about 5% of the marketed drugs suggesting their potential in drug discovery. The current review summarizes the therapeutic management of the principal ion channelopathies of central and peripheral nervous system, heart, kidney, bone, skeletal muscle and pancreas, resulting from mutations in calcium, sodium, potassium, and chloride ion channels. For most channelopathies the therapy is mainly empirical and symptomatic, often limited by lack of efficacy and tolerability for a significant number of patients. Other channelopathies can exploit ion channel targeted drugs, such as marketed sodium channel blockers. Developing new and more specific therapeutic approaches is therefore required. To this aim, a major advancement in the pharmacotherapy of channelopathies has been the discovery that ion channel mutations lead to change in biophysics that can in turn specifically modify the sensitivity to drugs: this opens the way to a pharmacogenetics strategy, allowing the development of a personalized therapy with increased efficacy and reduced side effects. In addition, the identification of disease modifiers in ion channelopathies appears an alternative strategy to discover novel druggable targets.
Collapse
Affiliation(s)
- Paola Imbrici
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| | - Antonella Liantonio
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| | - Giulia M Camerino
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| | - Michela De Bellis
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| | - Claudia Camerino
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro" Bari, Italy
| | - Antonietta Mele
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| | - Arcangela Giustino
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro" Bari, Italy
| | - Sabata Pierno
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| | - Annamaria De Luca
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| | - Domenico Tricarico
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| | - Jean-Francois Desaphy
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro" Bari, Italy
| | - Diana Conte
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro" Bari, Italy
| |
Collapse
|
81
|
Expression of a Diverse Array of Ca2+-Activated K+ Channels (SK1/3, IK1, BK) that Functionally Couple to the Mechanosensitive TRPV4 Channel in the Collecting Duct System of Kidney. PLoS One 2016; 11:e0155006. [PMID: 27159616 PMCID: PMC4861333 DOI: 10.1371/journal.pone.0155006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/22/2016] [Indexed: 12/02/2022] Open
Abstract
The voltage- and Ca2+-activated, large conductance K+ channel (BK, maxi-K) is expressed in the collecting duct system of kidney where it underlies flow- and Ca2+-dependent K+ excretion. To determine if other Ca2+-activated K+ channels (KCa) may participate in this process, mouse kidney and the K+-secreting mouse cortical collecting duct (CCD) cell line, mCCDcl1, were assessed for TRPV4 and KCa channel expression and cross-talk. qPCR mRNA analysis and immunocytochemical staining demonstrated TRPV4 and KCa expression in mCCDcl1 cells and kidney connecting tubule (CNT) and CCD. Three subfamilies of KCa channels were revealed: the high Ca2+-binding affinity small-conductance SK channels, SK1and SK3, the intermediate conductance channel, IK1, and the low Ca2+-binding affinity, BK channel (BKα subunit). Apparent expression levels varied in CNT/CCD where analysis of CCD principal cells (PC) and intercalated cells (IC) demonstrated differential staining: SK1:PC<IC, and SK3:PC>IC, IK1:PC>IC, BKα:PC = IC, and TRPV4:PC>IC. Patch clamp analysis and fluorescence Ca2+ imaging of mCCDcl1 cells demonstrated potent TRPV4-mediated Ca2+ entry and strong functional cross-talk between TRPV4 and KCa channels. TRPV4-mediated Ca2+ influx activated each KCa channel, as evidenced by selective inhibition of KCa channels, with each active KCa channel enhancing Ca2+ entry (due to membrane hyperpolarization). Transepithelial electrical resistance (TEER) analysis of confluent mCCDcl1 cells grown on permeable supports further demonstrated this cross-talk where TRPV4 activation induce a decrease in TEER which was partially restored upon selective inhibition of each KCa channel. It is concluded that SK1/SK3 and IK1 are highly expressed along with BKα in CNT and CCD and are closely coupled to TRPV4 activation as observed in mCCDcl1 cells. The data support a model in CNT/CCD segments where strong cross talk between TRPV4-mediated Ca2+ influx and each KCa channel leads to enhance Ca2+ entry which will support activation of the low Ca2+-binding affinity BK channel to promote BK-mediated K+ secretion.
Collapse
|
82
|
|
83
|
Nakamura T, Arima-Yoshida F, Sakaue F, Nasu-Nishimura Y, Takeda Y, Matsuura K, Akshoomoff N, Mattson SN, Grossfeld PD, Manabe T, Akiyama T. PX-RICS-deficient mice mimic autism spectrum disorder in Jacobsen syndrome through impaired GABAA receptor trafficking. Nat Commun 2016; 7:10861. [PMID: 26979507 PMCID: PMC4799364 DOI: 10.1038/ncomms10861] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 01/27/2016] [Indexed: 11/09/2022] Open
Abstract
Jacobsen syndrome (JBS) is a rare congenital disorder caused by a terminal deletion of the long arm of chromosome 11. A subset of patients exhibit social behavioural problems that meet the diagnostic criteria for autism spectrum disorder (ASD); however, the underlying molecular pathogenesis remains poorly understood. PX-RICS is located in the chromosomal region commonly deleted in JBS patients with autistic-like behaviour. Here we report that PX-RICS-deficient mice exhibit ASD-like social behaviours and ASD-related comorbidities. PX-RICS-deficient neurons show reduced surface γ-aminobutyric acid type A receptor (GABAAR) levels and impaired GABAAR-mediated synaptic transmission. PX-RICS, GABARAP and 14-3-3ζ/θ form an adaptor complex that interconnects GABAAR and dynein/dynactin, thereby facilitating GABAAR surface expression. ASD-like behavioural abnormalities in PX-RICS-deficient mice are ameliorated by enhancing inhibitory synaptic transmission with a GABAAR agonist. Our findings demonstrate a critical role of PX-RICS in cognition and suggest a causal link between PX-RICS deletion and ASD-like behaviour in JBS patients.
Collapse
Affiliation(s)
- Tsutomu Nakamura
- Laboratory of Molecular and Genetic Information, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Fumiko Arima-Yoshida
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Fumika Sakaue
- Laboratory of Molecular and Genetic Information, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Yukiko Nasu-Nishimura
- Laboratory of Molecular and Genetic Information, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Yasuko Takeda
- Laboratory of Molecular and Genetic Information, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Ken Matsuura
- Laboratory of Molecular and Genetic Information, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Natacha Akshoomoff
- Department of Psychiatry, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Sarah N Mattson
- Department of Psychology, San Diego State University, San Diego, California 92120, USA
| | - Paul D Grossfeld
- Department of Pediatrics, School of Medicine, University of California, San Diego, San Diego, California 92123, USA
| | - Toshiya Manabe
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Tetsu Akiyama
- Laboratory of Molecular and Genetic Information, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan
| |
Collapse
|
84
|
Affiliation(s)
- Sujay V Kharade
- a Department of Anesthesiology ; Vanderbilt University Medical Center ; Nashville , TN USA
| | | | | |
Collapse
|
85
|
Swale DR, Sheehan JH, Banerjee S, Husni AS, Nguyen TT, Meiler J, Denton JS. Computational and functional analyses of a small-molecule binding site in ROMK. Biophys J 2016; 108:1094-103. [PMID: 25762321 DOI: 10.1016/j.bpj.2015.01.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/21/2015] [Accepted: 01/23/2015] [Indexed: 12/20/2022] Open
Abstract
The renal outer medullary potassium channel (ROMK, or Kir1.1, encoded by KCNJ1) critically regulates renal tubule electrolyte and water transport and hence blood volume and pressure. The discovery of loss-of-function mutations in KCNJ1 underlying renal salt and water wasting and lower blood pressure has sparked interest in developing new classes of antihypertensive diuretics targeting ROMK. The recent development of nanomolar-affinity small-molecule inhibitors of ROMK creates opportunities for exploring the chemical and physical basis of ligand-channel interactions required for selective ROMK inhibition. We previously reported that the bis-nitro-phenyl ROMK inhibitor VU591 exhibits voltage-dependent knock-off at hyperpolarizing potentials, suggesting that the binding site is located within the ion-conduction pore. In this study, comparative molecular modeling and in silico ligand docking were used to interrogate the full-length ROMK pore for energetically favorable VU591 binding sites. Cluster analysis of 2498 low-energy poses resulting from 9900 Monte Carlo docking trajectories on each of 10 conformationally distinct ROMK comparative homology models identified two putative binding sites in the transmembrane pore that were subsequently tested for a role in VU591-dependent inhibition using site-directed mutagenesis and patch-clamp electrophysiology. Introduction of mutations into the lower site had no effect on the sensitivity of the channel to VU591. In contrast, mutations of Val(168) or Asn(171) in the upper site, which are unique to ROMK within the Kir channel family, led to a dramatic reduction in VU591 sensitivity. This study highlights the utility of computational modeling for defining ligand-ROMK interactions and proposes a mechanism for inhibition of ROMK.
Collapse
Affiliation(s)
- Daniel R Swale
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan H Sheehan
- Center for Structural Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sreedatta Banerjee
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Afeef S Husni
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Thuy T Nguyen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jens Meiler
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Structural Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee; Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Institute for Global Health, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
86
|
Stankewich MC, Moeckel GW, Ji L, Ardito T, Morrow JS. Isoforms of Spectrin and Ankyrin Reflect the Functional Topography of the Mouse Kidney. PLoS One 2016; 11:e0142687. [PMID: 26727517 PMCID: PMC4703142 DOI: 10.1371/journal.pone.0142687] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 10/26/2015] [Indexed: 11/24/2022] Open
Abstract
The kidney displays specialized regions devoted to filtration, selective reabsorption, and electrolyte and metabolite trafficking. The polarized membrane pumps, channels, and transporters responsible for these functions have been exhaustively studied. Less examined are the contributions of spectrin and its adapter ankyrin to this exquisite functional topography, despite their established contributions in other tissues to cellular organization. We have examined in the rodent kidney the expression and distribution of all spectrins and ankyrins by qPCR, Western blotting, immunofluorescent and immuno electron microscopy. Four of the seven spectrins (αΙΙ, βΙ, βΙΙ, and βΙΙΙ) are expressed in the kidney, as are two of the three ankyrins (G and B). The levels and distribution of these proteins vary widely over the nephron. αΙΙ/βΙΙ is the most abundant spectrin, found in glomerular endothelial cells; on the basolateral membrane and cytoplasmic vesicles in proximal tubule cells and in the thick ascending loop of Henle; and less so in the distal nephron. βΙΙΙ spectrin largely replaces βΙΙ spectrin in podocytes, Bowman’s capsule, and throughout the distal tubule and collecting ducts. βΙ spectrin is only marginally expressed; its low abundance hinders a reliable determination of its distribution. Ankyrin G is the most abundant ankyrin, found in capillary endothelial cells and all tubular segments. Ankyrin B populates Bowman’s capsule, podocytes, the ascending thick loop of Henle, and the distal convoluted tubule. Comparison to the distribution of renal protein 4.1 isoforms and various membrane proteins indicates a complex relationship between the spectrin scaffold, its adapters, and various membrane proteins. While some proteins (e.g. ankyrin B, βΙΙΙ spectrin, and aquaporin 2) tend to share a similar distribution, there is no simple mapping of different spectrins or ankyrins to most membrane proteins. The implications of this data are discussed.
Collapse
Affiliation(s)
- Michael C. Stankewich
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
- * E-mail:
| | - Gilbert W. Moeckel
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
| | - Lan Ji
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
| | - Thomas Ardito
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
| | - Jon S. Morrow
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States of America
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, United States of America
| |
Collapse
|
87
|
Chapter Five - Ubiquitination of Ion Channels and Transporters. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:161-223. [DOI: 10.1016/bs.pmbts.2016.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
88
|
Abstract
More than two dozen types of potassium channels, with different biophysical and regulatory properties, are expressed in the kidney, influencing renal function in many important ways. Recently, a confluence of discoveries in areas from human genetics to physiology, cell biology, and biophysics has cast light on the special function of five different potassium channels in the distal nephron, encoded by the genes KCNJ1, KCNJ10, KCNJ16, KCNMA1, and KCNN3. Research aimed at understanding how these channels work in health and go awry in disease has transformed our understanding of potassium balance and provided new insights into mechanisms of renal sodium handling and the maintenance of blood pressure. This review focuses on recent advances in this rapidly evolving field.
Collapse
Affiliation(s)
- Paul A Welling
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201;
| |
Collapse
|
89
|
Pilmore E, Hamilton KL. The Role of MicroRNAs in the Regulation of K(+) Channels in Epithelial Tissue. Front Physiol 2015; 6:352. [PMID: 26648872 PMCID: PMC4664832 DOI: 10.3389/fphys.2015.00352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/09/2015] [Indexed: 12/27/2022] Open
Abstract
Our understanding of the modulation of proteins has shifted in direction with the discovery of microRNAs (miRs) over twenty years ago. MiRs are now in the “limelight” as these non-coding pieces of RNA (generally ~22 nucleotides long) result in altered translation and function of proteins. Indeed, miRs are now reported to be potential biomarkers of disease. Epithelial K+ channels play many roles in electrolyte and fluid homeostasis of the human body and have been suggested to be therapeutic targets of disease. Interestingly, the role of miRs in modulating K+ channels of epithelial tissues is only emerging now. This minireview focuses on recent novel findings into the role of miRs in the regulation of K+ channels of epithelia.
Collapse
Affiliation(s)
- Elliot Pilmore
- Department of Physiology, Otago School of Medical Sciences, University of Otago Dunedin, New Zealand
| | - Kirk L Hamilton
- Department of Physiology, Otago School of Medical Sciences, University of Otago Dunedin, New Zealand
| |
Collapse
|
90
|
Blass B. Inhibitors of the Renal Outer Medullary Potassium Channel. ACS Med Chem Lett 2015; 6:831-3. [PMID: 26288678 DOI: 10.1021/acsmedchemlett.5b00227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Indexed: 11/28/2022] Open
Affiliation(s)
- Benjamin Blass
- Temple University School of Pharmacy , 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
91
|
Wu Y, Baum M, Huang CL, Rodan AR. Two inwardly rectifying potassium channels, Irk1 and Irk2, play redundant roles in Drosophila renal tubule function. Am J Physiol Regul Integr Comp Physiol 2015. [PMID: 26224687 DOI: 10.1152/ajpregu.00148.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Inwardly rectifying potassium channels play essential roles in renal physiology across phyla. Barium-sensitive K(+) conductances are found on the basolateral membrane of a variety of insect Malpighian (renal) tubules, including Drosophila melanogaster. We found that barium decreases the lumen-positive transepithelial potential difference in isolated perfused Drosophila tubules and decreases fluid secretion and transepithelial K(+) flux. In those insect species in which it has been studied, transcripts from multiple genes encoding inwardly rectifying K(+) channels are expressed in the renal (Malpighian) tubule. In Drosophila melanogaster, this includes transcripts of the Irk1, Irk2, and Irk3 genes. The role of each of these gene products in renal tubule function is unknown. We found that simultaneous knockdown of Irk1 and Irk2 in the principal cell of the fly tubule decreases transepithelial K(+) flux, with no additive effect of Irk3 knockdown, and decreases barium sensitivity of transepithelial K(+) flux by ∼50%. Knockdown of any of the three inwardly rectifying K(+) channels individually has no effect, nor does knocking down Irk3 simultaneously with Irk1 or Irk2. Irk1/Irk2 principal cell double-knockdown tubules remain sensitive to the kaliuretic effect of cAMP. Inhibition of the Na(+)/K(+)-ATPase with ouabain and Irk1/Irk2 double knockdown have additive effects on K(+) flux, and 75% of transepithelial K(+) transport is due to Irk1/Irk2 or ouabain-sensitive pathways. In conclusion, Irk1 and Irk2 play redundant roles in transepithelial ion transport in the Drosophila melanogaster renal tubule and are additive to Na(+)/K(+)-ATPase-dependent pathways.
Collapse
Affiliation(s)
- Yipin Wu
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Michel Baum
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas; and Department of Pediatrics, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chou-Long Huang
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Aylin R Rodan
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| |
Collapse
|
92
|
Raymer B, Ebner D. Small molecule and peptide therapies for chronic heart failure: a patent review (2011 - 2014). Expert Opin Ther Pat 2015; 25:1175-90. [PMID: 26173447 DOI: 10.1517/13543776.2015.1061997] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Chronic heart failure (CHF) is the long-term inability of the heart to meet circulatory demands under normal conditions. Effects of CHF can include increased blood volume, increased vascular resistance and compromised contractility leading to fluid retention, dyspnea and fatigue. Current standard of care for chronic systolic heart failure is directed towards managing hypoperfusion through the renin-angiotensin-aldosterone and sympathetic nervous systems. Treatment may also involve reversal of maladaptive cardiac remodeling and prevention of life-threatening arrhythmias. AREAS COVERED This review highlights small molecule and peptidic agents for the treatment of CHF with patents published between 2011 and 2014. Targets are subdivided into inotropic agents, ventricular remodeling, diuretics and the renin-angiotensin-aldosterone system. EXPERT OPINION CHF represents a large, unmet medical need where improved therapies are needed. The renin-angiotensin-aldosterone system pathway continues to be a major source of new therapies for CHF with new inotropic therapies emerging. Promising initial clinical results for a few approaches combined with the expectation of additional clinical results in the near future make this an exciting time in the pursuit of new treatments for CHF.
Collapse
Affiliation(s)
- Brian Raymer
- a Cardiovascular, Metabolic, and Endocrine Diseases Chemistry, Pfizer Worldwide Research and Development , Cambridge, MA, USA +1 617 551 3414 ; +1 617 551 3082 ;
| | - David Ebner
- a Cardiovascular, Metabolic, and Endocrine Diseases Chemistry, Pfizer Worldwide Research and Development , Cambridge, MA, USA +1 617 551 3414 ; +1 617 551 3082 ;
| |
Collapse
|
93
|
Guh YJ, Lin CH, Hwang PP. Osmoregulation in zebrafish: ion transport mechanisms and functional regulation. EXCLI JOURNAL 2015; 14:627-59. [PMID: 26600749 PMCID: PMC4650948 DOI: 10.17179/excli2015-246] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/21/2015] [Indexed: 12/21/2022]
Abstract
Fish, like mammals, have to maintain their body fluid ionic and osmotic homeostasis through sophisticated iono-/osmoregulation mechanisms, which are conducted mainly by ionocytes of the gill (the skin in embryonic stages), instead of the renal tubular cells in mammals. Given the advantages in terms of genetic database availability and manipulation, zebrafish is an emerging model for research into regulatory and integrative physiology. At least five types of ionocytes, HR, NaR, NCC, SLC26, and KS cells, have been identified to carry out Na(+) uptake/H(+) secretion/NH4 (+) excretion, Ca(2+) uptake, Na(+)/Cl(-) uptake, K(+) secretion, and Cl(-) uptake/HCO3 (-) secretion, respectively, through distinct sets of transporters. Several hormones, namely isotocin, prolactin, cortisol, stanniocalcin-1, calcitonin, endothelin-1, vitamin D, parathyorid hormone 1, catecholamines, and the renin-angiotensin-system, have been demonstrated to positively or negatively regulate ion transport through specific receptors at different ionocytes stages, at either the transcriptional/translational or posttranslational level. The knowledge obtained using zebrafish answered many long-term contentious or unknown issues in the field of fish iono-/osmoregulation. The homology of ion transport pathways and hormone systems also means that the zebrafish model informs studies on mammals or other animal species, thereby providing insights into related fields.
Collapse
Affiliation(s)
- Ying-Jey Guh
- Institute of Cellular and Organismic Biology, Academia Sinica, Nakang, Taipei, Taiwan ; Institute of Biological Chemistry, Academia Sinica, Nakang, Taipei, Taiwan
| | - Chia-Hao Lin
- National Institute for Basic Biology, Myodaiji-cho, Okazaki, 444-8787, Japan
| | - Pung-Pung Hwang
- Institute of Cellular and Organismic Biology, Academia Sinica, Nakang, Taipei, Taiwan
| |
Collapse
|
94
|
Horn S, Naidus E, Alper SL, Danziger J. Cesium-associated hypokalemia successfully treated with amiloride. Clin Kidney J 2015; 8:335-8. [PMID: 26034598 PMCID: PMC4440464 DOI: 10.1093/ckj/sfv017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/26/2015] [Indexed: 12/12/2022] Open
Abstract
Self-treatment of cancer with cesium chloride, despite proven lack of efficacy, continues to produce serious adverse effects. Among these is hypokalemia predisposing to life-threatening arrhythmia. The mechanism of cesium-associated hypokalemia (CAH) has not been described. We report urinary potassium wasting responsive to amiloride therapy in a cancer patient with CAH, and discuss possible mechanisms.
Collapse
Affiliation(s)
- Sarah Horn
- Beth Israel Deaconess Medical Center , Harvard Medical School , Boston, MA , USA
| | - Elliot Naidus
- Beth Israel Deaconess Medical Center , Harvard Medical School , Boston, MA , USA
| | - Seth L Alper
- Beth Israel Deaconess Medical Center , Harvard Medical School , Boston, MA , USA
| | - John Danziger
- Beth Israel Deaconess Medical Center , Harvard Medical School , Boston, MA , USA
| |
Collapse
|
95
|
Abstract
The amiloride-sensitive epithelial Na(+) channel (ENaC) is a key player in the regulation of Na(+) homeostasis. Its functional activity is under continuous control by a variety of signaling molecules, including bioactive peptides of endothelin family. Since ENaC dysfunction is causative for disturbances in total body Na(+) levels associated with the abnormal regulation of blood volume, blood pressure, and lung fluid balance, uncovering the molecular mechanisms of inhibitory modulation or inappropriate activation of ENaC is crucial for the successful treatment of a variety of human diseases including hypertension. The precise regulation of ENaC is particularly important for normal Na(+) and fluid homeostasis in organs where endothelins are known to act: the kidneys, lung, and colon. Inhibition of ENaC by endothelin-1 (ET-1) has been established in renal cells, and several molecular mechanisms of inhibition of ENaC by ET-1 are proposed and will be reviewed in this chapter.
Collapse
Affiliation(s)
- Andrey Sorokin
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| | | |
Collapse
|
96
|
Sepúlveda FV, Pablo Cid L, Teulon J, Niemeyer MI. Molecular aspects of structure, gating, and physiology of pH-sensitive background K2P and Kir K+-transport channels. Physiol Rev 2015; 95:179-217. [PMID: 25540142 DOI: 10.1152/physrev.00016.2014] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
K(+) channels fulfill roles spanning from the control of excitability to the regulation of transepithelial transport. Here we review two groups of K(+) channels, pH-regulated K2P channels and the transport group of Kir channels. After considering advances in the molecular aspects of their gating based on structural and functional studies, we examine their participation in certain chosen physiological and pathophysiological scenarios. Crystal structures of K2P and Kir channels reveal rather unique features with important consequences for the gating mechanisms. Important tasks of these channels are discussed in kidney physiology and disease, K(+) homeostasis in the brain by Kir channel-equipped glia, and central functions in the hearing mechanism in the inner ear and in acid secretion by parietal cells in the stomach. K2P channels fulfill a crucial part in central chemoreception probably by virtue of their pH sensitivity and are central to adrenal secretion of aldosterone. Finally, some unorthodox behaviors of the selectivity filters of K2P channels might explain their normal and pathological functions. Although a great deal has been learned about structure, molecular details of gating, and physiological functions of K2P and Kir K(+)-transport channels, this has been only scratching at the surface. More molecular and animal studies are clearly needed to deepen our knowledge.
Collapse
Affiliation(s)
- Francisco V Sepúlveda
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - L Pablo Cid
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - Jacques Teulon
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| | - María Isabel Niemeyer
- Centro de Estudios Científicos, Valdivia, Chile; UPMC Université Paris 06, Team 3, Paris, France; and Institut National de la Santé et de la Recherche Médicale, UMR_S 1138, Paris, France
| |
Collapse
|
97
|
Affiliation(s)
- Lonny R. Levin
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065; ,
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065; ,
| |
Collapse
|
98
|
Abstract
The distal convoluted tubule (DCT) is a short nephron segment, interposed between the macula densa and collecting duct. Even though it is short, it plays a key role in regulating extracellular fluid volume and electrolyte homeostasis. DCT cells are rich in mitochondria, and possess the highest density of Na+/K+-ATPase along the nephron, where it is expressed on the highly amplified basolateral membranes. DCT cells are largely water impermeable, and reabsorb sodium and chloride across the apical membrane via electroneurtral pathways. Prominent among this is the thiazide-sensitive sodium chloride cotransporter, target of widely used diuretic drugs. These cells also play a key role in magnesium reabsorption, which occurs predominantly, via a transient receptor potential channel (TRPM6). Human genetic diseases in which DCT function is perturbed have provided critical insights into the physiological role of the DCT, and how transport is regulated. These include Familial Hyperkalemic Hypertension, the salt-wasting diseases Gitelman syndrome and EAST syndrome, and hereditary hypomagnesemias. The DCT is also established as an important target for the hormones angiotensin II and aldosterone; it also appears to respond to sympathetic-nerve stimulation and changes in plasma potassium. Here, we discuss what is currently known about DCT physiology. Early studies that determined transport rates of ions by the DCT are described, as are the channels and transporters expressed along the DCT with the advent of molecular cloning. Regulation of expression and activity of these channels and transporters is also described; particular emphasis is placed on the contribution of genetic forms of DCT dysregulation to our understanding.
Collapse
Affiliation(s)
- James A McCormick
- Division of Nephrology & Hypertension, Oregon Health & Science University, & VA Medical Center, Portland, Oregon, United States
| | | |
Collapse
|
99
|
Furukawa F, Watanabe S, Kakumura K, Hiroi J, Kaneko T. Gene expression and cellular localization of ROMKs in the gills and kidney of Mozambique tilapia acclimated to fresh water with high potassium concentration. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1303-12. [PMID: 25298512 DOI: 10.1152/ajpregu.00071.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regulation of plasma K(+) levels in narrow ranges is vital to vertebrate animals. Since seawater (SW) teleosts are loaded with excess K(+), they constantly excrete K(+) from the gills. However, the K(+) regulatory mechanisms in freshwater (FW)-acclimated teleosts are still unclear. We aimed to identify the possible K(+) regulatory mechanisms in the gills and kidney, the two major osmoregulatory organs, of FW-acclimated Mozambique tilapia (Oreochromis mossambicus). As a potential molecular candidate for renal K(+) handling, a putative renal outer medullary K(+) channel (ROMK) was cloned from the tilapia kidney and tentatively named "ROMKb"; another ROMK previously cloned from the tilapia gills was thus renamed "ROMKa". The fish were acclimated to control FW or to high-K(+) (H-K) FW for 1 wk, and we assessed physiological responses of tilapia to H-K treatment. As a result, urinary K(+) levels were slightly higher in H-K fish, implying a role of the kidney in K(+) excretion. However, the mRNA expression levels of both ROMKa and ROMKb were very low in the kidney, while that of K(+)/Cl(-) cotransporter 1 (KCC1) was robust. In the gills, ROMKa mRNA was markedly upregulated in H-K fish. Immunofluorescence staining showed that branchial ROMKa was expressed at the apical membrane of type I and type III ionocytes, and the ROMKa immunosignals were more intense in H-K fish than in control fish. The present study suggests that branchial ROMKa takes a central role for K(+) regulation in FW conditions and that K(+) excretion via the gills is activated irrespective of environmental salinity.
Collapse
Affiliation(s)
- Fumiya Furukawa
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan; and
| | - Soichi Watanabe
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan; and
| | - Keigo Kakumura
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan; and
| | - Junya Hiroi
- Department of Anatomy, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Toyoji Kaneko
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan; and
| |
Collapse
|
100
|
Kamel KS, Schreiber M, Halperin ML. Integration of the response to a dietary potassium load: a paleolithic perspective. Nephrol Dial Transplant 2014; 29:982-9. [PMID: 24789504 DOI: 10.1093/ndt/gft499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Our purpose is to integrate new insights in potassium (K(+)) physiology to understand K(+) homeostasis and illustrate some of their clinical implications. Since control mechanisms that are essential for survival were likely developed in Paleolithic times, we think the physiology of K(+) homeostasis can be better revealed when viewed from what was required to avoid threats and achieve balance in Paleolithic times. Three issues will be highlighted. First, we shall consider the integrative physiology of the gastrointestinal tract and the role of lactic acid released from enterocytes following absorption of sugars (fruit and berries) to cause a shift of this K(+) load into the liver. Second, we shall discuss the integrative physiology of WNK kinases and modulation of delivery of bicarbonate to the distal nephron to switch the aldosterone response from sodium chloride retention to K(+) secretion when faced with a K(+) load. Third, we shall emphasize the role of intra-renal recycling of urea in achieving K(+) homeostasis when the diet contains protein and K(+).
Collapse
Affiliation(s)
- Kamel S Kamel
- Renal Division, St Michael's Hospital and University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|